You are on page 1of 10

1364 DOI 10.1002/mnfr.201500955 Mol. Nutr. Food Res.

2016, 60, 1364–1373

REVIEW

Ginger augmented chemotherapy: A novel multitarget


nontoxic approach for cancer management
Roopali Saxena1 , Padmashree C. G. Rida1 , Omer Kucuk2 and Ritu Aneja1
1
Department of Biology, Georgia State University, Atlanta, GA, USA
2
Winship Cancer Institute of Emory University, Atlanta, GA, USA

Cancer, referred to as the ‘disease of civilization’, continues to haunt humanity due to its dread- Received: November 24, 2015
ful manifestations and limited success of therapeutic interventions such as chemotherapy in Revised: January 11, 2016
curing the disease. Although effective, chemotherapy has repeatedly demonstrated inadequacy Accepted: January 12, 2016
in disease management due to its debilitating side effects arising from its deleterious non-
specific effects on normal healthy cells. In addition, development of chemoresistance due to
mono-targeting often results in cessation of chemotherapy. This urgently demands develop-
ment and implementation of multitargeted alternative therapies with mild or no side effects.
One extremely promising strategy that yet remains untapped in the clinic is augmenting
chemotherapy with dietary phytochemicals or extracts. Ginger, depository of numerous bioac-
tive molecules, not only targets cancer cells but can also mitigate chemotherapy-associated
side effects. Consequently, combination therapy involving ginger extract and chemotherapeu-
tic agents may offer the advantage of being efficacious with reduced toxicity. Here we discuss
the remarkable and often overlooked potential of ginger extract to manage cancer, the possibil-
ity of developing ginger-based combinational therapies, and the major roadblocks along with
strategies to overcome them in clinical translation of such inventions. We are optimistic that
clinical implementation of such combination regimens would be a much sought after modality
in cancer management.

Keywords:
Antiemetic / Cancer / Chemotherapy / Ginger / Gingerols / Shogaols

1 Introduction ramifications on society with global spending on cancer


medication reaching 100 billion dollars in 2014 [1]. There-
Cancer represents one of the deadliest and fastest growing ail- fore, speedy increase in cancer incidence and economic
ments worldwide with soaring morbidity and mortality. Ac- burden due to cost of cancer care present an alarming
cording to GLOBOCAN 2012, an epidemiological database situation for future and demand an immediate attention
for cancer prevalence worldwide, approximately 14 million to revisit conventional strategies for disease prevention
new cases were reported in 2012 alone, along with 8.2 mil- and cure.
lion cancer-related deaths. It is projected that reported annual Cancer is a heterogeneous and constantly evolving disease
new cases would rise to a staggering 22 million over the next with genetic and epigenetic changes occurring in the cancer
two decades (WHO, GLOBOCAN). Cancer not only inflicts cells as well as structural and architectural changes ensuing
damage on human life but also has far-reaching economic in the microenvironment. At the cellular level, cancer may
be defined as a mass of phenotypically and genotypically dis-
similar abnormal cells that defy the rules of conventional cell
Correspondence: Ritu Aneja
E-mail: raneja@gsu.edu proliferation and death pathways. In essence, cancer is a col-
lective term encompassing a conglomerate of diseases that
Abbreviations: Cdks, Cyclin-dependent kinases; CINV, present with specific hallmarks. As a result, cancer cells ac-
Chemotherapy-induced nausea and vomiting; CPT11, Irinotecan; quire a number of characteristics over time that confer them
6DG, 6-Dehydrogingerdione; ERK, Extracellular signal-regulated
with the ability to divide erratically and give them the ad-
kinases; GE, Ginger extract; 6G, [6]-Gingerol; 8G, [8]-Gingerol;
vantage to invade other parts of the body. These hallmarks
10G, [10]-Gingerol; 5-HT3 , 5-Hydroxytryptamine 3; JNK, c-Jun N-
terminal kinase; MAPK, Mitogen-activated protein kinase; NK-1, include but are not limited to, sustained cell proliferation,
Neurokinin-1; ROS, Reactive oxygen species; 6S, [6]-Shogaol;
4S, [4]-Shogaol Colour Online: See the article online to view Figs. 1 and 2 in colour.


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2016, 60, 1364–1373 1365

avoidance of resistance to cell death, induction of angiogene- chemotherapeutic agents used for cancer treatment that can
sis, and invasion of normal healthy cells [2]. Although cancer be categorized into cytotoxic drugs (anti-metabolites, alkylat-
formation is a multifactorial phenomenon, there is enough ing agents, plant derivatives), hormones (agonists and antag-
evidence that underlines the importance of lifestyle factors onists) and other agents (monoclonal antibodies) [9]. These
in fueling carcinogenesis [3]. These factors include reduced cytotoxic agents were strategically designed to kill cells with
intake of fruits and vegetables, lack of exercise, stress, obe- high proliferation rate, which was believed to be forte of can-
sity, use of tobacco etc. A combination of these factors, in cer cells. As a result, these drugs target other rapidly diving
association with environmental elements, leads to chronic healthy cells in the body resulting in the manifestation of ad-
inflammation and oxidative stress, primary culprits respon- verse side effects of gastrointestinal, respiratory, cardiac, hep-
sible for the malignant transformation of cells. atic and neurological nature (Fig. 1A). Majority of currently
Growing incidence of cancer and escalating healthcare used chemotherapeutic agents target either DNA or various
costs have sparked interest in the development of chemo- steps of cell division/mitosis. For instance, taxanes such as
preventive measures which inhibit and disrupt cancer de- paclitaxel or docetaxel disrupt microtubules assembly which
velopment and progression through long-term use of syn- is required for proper cell division [10], alkylating agents such
thetic, natural, or biological agents with minimal or no side as cyclophosphamide damage DNA leading to apoptosis [11],
effects [4]. Owing to genotypic and phenotypic intratumor platinum compounds such as cisplatin interfere with cell di-
heterogeneity, an ideal chemopreventive regimen should en- vision by crosslinking DNA strands [12] and topoisomerase
tail targeting multiple cell survival pathways in order to inhibitors like irinotecan target topoisomerases, which are
achieve utmost benefits. Dietary phytochemicals qualify as enzymes that regulate DNA topology during DNA replica-
excellent chemopreventive candidates due to their pleiotropic tion and transcription [13]. One of the underlying causes of
and nontoxic attributes. Numerous studies highlight the adverse side effects of these chemotherapeutic agents is ac-
role of dietary and lifestyle factors, which can regulate pro- companying death of noncancerous healthy cells with high
inflammatory pathways and oxidative damage to cellular proliferation rate. Cytotoxic effects of microtubule binding
macromolecules such as DNA and protein that lead to initia- agents on normal cells dwelling in interphase of cell cycle
tion of tumorigenesis [5]. Dietary bioactive compounds have further contribute to the toxicity of chemotherapy [14]. The
also been shown to regulate the activity of enzymes such as side effects of chemotherapy are classified as acute or late,
histone acetyl transferases and histone deacetylases respon- based on the time frame of their occurrence after therapy
sible for epigenetic changes whose accumulation over time [15]. Acute toxicity occurs within a few weeks of treatment
and other alterations in cellular homeostasis affect genome and affects tissues with high proliferation rate such as red
stability that can potentially cause carcinogenesis. [6]. In ad- blood cells of bone marrow, gastrointestinal lining, and skin.
dition, dietary phytochemicals have been reported to target On the other hand, late toxicity affects patients several months
a variety of signaling pathways whose dysregulation is im- post treatment and is mainly associated with tissues with low
plicated in cancer development such as MAPK/ERK and proliferation rate [15]. Some of the manifestations of late tox-
PI3K/Akt/mTOR pathways [7]. Some of these bioactive com- icity include neural and vascular damage, fibrosis, infertility,
pounds with significance in cancer prevention and manage- and secondary malignancy. Chemotherapy-induced nausea
ment include gingerols, shogaols, genistein, phenylisothio- and vomiting (CINV) is the most common and debilitating
cyanate, curcumin, resveratrol and indole-3-carbinol [6, 8]. side effect of chemotherapy caused by systemic cytotoxic-
Currently, combination therapy combining attributes of ity to gastrointestinal lining (Fig. 1B). It has been estimated
chemotherapy, radiation and immunotherapy is considered that approximately 70% of patients experience CINV despite
the most effective and prevalent approach for cancer treat- widespread use of antiemetic agents [16]. Chemotherapeu-
ment. Early stage detection of cancer often results in good tic agents are classified into four risk groups: high, mod-
survival rate, yet the effectiveness of combination therapies erate, low, and minimal depending on the severity of CINV
in disease management is significantly limited due to their upon their administration in the absence of antiemetic agents
associated side effects, which often result in discontinuation [17,18]. Drugs like cisplatin and streptozotocin falls under the
of lengthy treatment regimens. Therefore, there is growing category of high emetic risk group while agents such as hor-
interest in the development of phytonutrient-based augmen- mones, gemcitabine, fluorouracil, etc. belong to low emetic
tation of cancer therapy that offers the advantage of being less risk group. The exact mechanism of CINV has not been com-
toxic and more economical. pletely elucidated but it is suggested that chemotherapeutic
agents induce nausea and vomiting by increasing serotonin
concentration in intestine and brain stem [19]. Serotonin is
2 Conventional chemotherapy: A curse in a neurotransmitter that mediates its physiological responses
disguise via activation of serotonin receptors [20]. Serotonin medi-
ated activation of vomiting center and/or chemoreceptor trig-
Since the approval of the first chemotherapeutic drug in ger zone located in brain, via central and peripheral neural
1949, thousands of compounds have been screened as po- processes, has been proposed to be predominantly respon-
tential anticancer agents. Currently, there are more than 200 sible for CINV (Fig. 1B). 5-hydroxytryptamine receptors-3


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
1366 R. Saxena et al. Mol. Nutr. Food Res. 2016, 60, 1364–1373

Figure 1. (A) Side effects of chemotherapy. Chemotherapy is one of the most prevalent interventions to treat cancer but its application
causes various adverse side effects in patients as depicted in the figure. These side effects can be gastrointestinal, cardiac, respiratory,
hepatic, or neurological and their extent and severity vary by person and therapeutic agent. (B) Chemotherapy-induced nausea and
vomiting. CINV is the most prevailing side effects of chemotherapy. CINV is induced by stimulation of vomiting center and/or chemoreceptor
trigger zone located in brain through serotonin-activated neural pathways. Although patients are prescribed antiemetic agents to alleviate
CINV, benefits of such agents are limited due to their associated toxicity.

(5-HT3 ) are known to particularly mediate acute nausea and nature that are associated with specific chemotherapeu-
vomiting while activation of neurokinin-1 receptors (NK-1) tic agents [9]. For instance, cardiovascular complications
by substance P is associated with delayed responses [15]. are more commonly associated with the use of anthra-
Agents that block these receptors, termed antiemetic recep- cyclines, 5-fluororacil, and cyclophosphamide and include
tor antagonists, are normally prescribed to patients under- cardiomyopathy and heart failure due to myocyte death and fi-
going chemotherapy in order to limit CINV [21]. Commonly brosis, arrhythmias, coronary ischemia, and vascular inflam-
prescribed antiemetic agents include dopamine receptor an- mation [9, 25]. Although generation of free radicals and con-
tagonists such as haloperidol and metoclopramide, 5-HT3 sequent cellular damage are key mechanistic players in the
receptor antagonists such as ondansetron, granistron, and development of cardiotoxicity, other proposed mechanisms
dolasetron and NK-1 antagonist aprepitant [22]. Although ef- include modulation of intracellular ATP production and car-
fective, some of these emetic agents can themselves cause diac topoisomerase enzyme activity [25]. Patients undergo-
side effects, which limit their use in clinical practice. For in- ing chemotherapy generally develop secondary respiratory
stance, use of haloperidol can lead to dystonia and sedation infections due to immunosuppressive effects of chemother-
in treated patients [22]. In addition to CINV, acute diarrhea apy [9]. Hepatic impairment constitutes an uncommon and
is another well-known and common gastrointestinal side ef- unpredictable side effect of systemic chemotherapy and may
fect particularly associated with the use of chemotherapeutic lead to manifestations from jaundice to acute hepatic fail-
agents such as 5-fluorouracil and irinotecan (CPT11). The ure. Chemotherapy mediates renal disease and electrolyte
toxicity of CPT11 is due to its metabolic transformation into a disturbances by its effects on glomerulus, tubules, intersti-
toxic metabolite called SN38, which can cause mucosal dam- tium, or the renal microvasculature leading to delayed re-
age leading to exudative processes along with malabsorption nal clearance of chemotherapeutic agents and their active
of water and electrolytes [23, 24]. metabolites [9]. Further, chemotherapy can cause injuries to
While gastrointestinal side effects constitute the most central and peripheral nervous system leading to the devel-
prevalent and widely recognized side effects, chemother- opment of sensory and motor neuropathies, transient asep-
apy induces a wide array of other systemic perturbations of tic meningitis, transverse myelitis, seizures, and permanent
cardiovascular, respiratory, hepatic, renal, and neurological leucoencephalopathy [9]. Other notable adverse neurotoxic


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2016, 60, 1364–1373 1367

conditions include mood changes and neurocognitive alter- results in enrichment of shogaols in dried or thermally treated
ations such as memory loss, impaired attention, and decline ginger as opposed to fresh ginger that has large quantities of
in concentration and processing speed which are mediated gingerols [31].
by plausible effects of chemotherapy on hippocampal func- Since ginger harbors numerous bioactive molecules with
tion [26]. Although neurological side effects are observed diverse pharmacological properties, it can be appropriately
across the whole spectrum of cytotoxic agents, the exact referred to as ‘natural panacea’ and can be employed to treat
mechanism underlying these effects remains elusive owing a variety of ailments. Given the diverse composition of gin-
to the use of multiple agents during combination therapy [26]. ger, its beneficial effects in various pathological conditions
Chemotherapy-induced neurotoxicity represents a relatively are by virtue of targeting an array of signaling pathways.
new field of investigation in clinical oncology and neurotox- Ginger and its constituents can prevent the buildup of ox-
icity management is generally overlooked in patients due to idative stress that can damage cellular macromolecules un-
lack of validated tests and standardized guidelines for the derlying various pathological conditions [32]. This is achieved
assessment of neurological symptoms including cognitive in a variety of ways such as scavenging free radicals, modu-
dysfunction. Although neuroimaging techniques provide a lating levels of antioxidant molecules (glutathione) and en-
detailed analysis of cognitive function by assessing the struc- zymes (superoxide dismutase, catalase), and modulating lev-
tural and functional effects of chemotherapeutic agents on els of phase I (cytochrome P450) and phase II (glutathione
brain, studies should be aimed at designing and validating S-transferase) drug metabolizing enzymes [33]. Many gin-
simple neurological tests for accurate and quick assessment ger constituents possess anti-inflammatory properties and
of cognitive dysfunction in cancer patients [27]. Despite ex- exert their effects by suppression of prostaglandin and
tensive research focused on understanding and combating leukotriene synthesis and pro-inflammatory cytokines such
chemotherapy-induced side effects, detailed molecular mech- as interleukin-1, tumor necrosis factor-␣, and interleukin-8
anisms underlying chemotherapy-induced side effects are not [33–35]. Ginger acts as a dual inhibitor of enzymes such as
completely known and for some of them, no treatment yet cyclooxygenase 1 and 2 (COX 1 and 2) and 5-lipoxygenase (5-
exists. LO) which are involved in the biosynthesis of inflammation
mediators such as prostaglandins and leukotrienes [36, 37].
As a result, extended use and high doses of ginger do not
3 Ginger: A rooty panacea cause toxicity often associated with the use of NSAIDs which
shifts arachidonic metabolism toward increased synthesis
Indigenous to Southeast Asia, ginger is valued for its un- of pro-inflammatory leukotrienes [35, 38]. The broad effect
matched medicinal properties since ancient time and has of ginger on inflammation can be attributed to its ability
spread worldwide by means of spice trade. Ginger is referred to regulate expression of NF-␬B, a master regulator of pro-
to as the universal remedy and constitutes an integral part inflammatory genes coding for chemokines and cytokines
of traditional Chinese and Indian Ayurvedic medicine. The [35, 39]. For instance, it has been shown that ginger extract
underground stem of ginger plant, called as rhizome, has inhibits expression of NF-␬B in activated synoviocytes [40].
been traditionally known to treat a range of gastrointestinal The anti-microbial properties of ginger are mediated through
disorders such as constipation, dyspepsia, gastritis, indiges- anti-bacterial and anti-fungal effects of gingerols as well as
tion, nausea, and vomiting along with other ailments includ- other bioactive constituents such as paradols, zingerone, and
ing arthritis, rheumatism, sprains, muscle aches, sore throat, shogaols [34]. Further, ginger constituents are reported to
cramps, hypertension, dementia, fever, gingivitis, asthma, modulate ‘tumor-permissive’ signaling pathways, which con-
stroke, and diabetes [8]. The phytochemical composition of tribute to the antitumor property of ginger. For example,
ginger rhizome is diverse containing both volatile and non- [6]-gingerol (6G) has been shown to block p38 MAP kinase-
volatile molecules, responsible for its flavor and aroma, along NF-␬B signaling pathway by suppressing the expression of
with a proteolytic enzyme called zingibin and the relative cyclooxygenase-2 [41, 42] while [6]-shogoal (6S) inhibited NF-
abundances of ginger constituents depend on various ge- ␬B activation by preventing dimerization of Toll-like receptors
ographical, environmental, and agricultural factors [28, 29]. 4, attesting to pleiotropic nature of ginger constituents [43].
The volatile component of ginger is mainly constituted of
mono- and sesquiterpenes with zingiberol being the prin-
cipal aroma-contributing molecule. The nonvolatile compo- 4 Ginger in the war against cancer:
nents are responsible for pungency of ginger and consist Where do we stand today?
mainly of gingerols, shogaols, paradols, and zingerone [30].
These compounds are biologically active and account for most The anticancer property of ginger needs no introduction
of the pharmacological effects of ginger [28]. Gingerols con- and is predominantly mediated by its constituents such as
stitute a series of chemical homologs that differ from each gingerols, shogaols, and paradols. Numerous studies con-
other in alkyl chain length and a side chain, with 6-gingerol ducted in the last two decades have demonstrated that ginger
being the most abundant one. Gingerols are sensitive to heat extract (GE) and its constituents exhibit antitumor activity
and can easily undergo dehydration to form shogaols. This against a variety of cancer cell lines and tumor xenografts by


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
1368 R. Saxena et al. Mol. Nutr. Food Res. 2016, 60, 1364–1373

employing multiple mechanisms [33]. 6G has been shown to [56]. Although further studies are warranted, we extrapolated
inhibit cell proliferation and induce apoptosis in human col- mice efficacy data to humans by performing allometric scal-
orectal cancer cells through downregulation of cyclin D1 and ing calculations, and the human equivalent effective dose
upregulation of pro-apoptotic cytokine called nonsteroidal of GE for reduction of tumor burden was found to be 567
anti-inflammatory drug (NSAID)-activated gene-1 [44]. On mg for a 70 kg adult [57]. Reports from randomized human
the other hand, [10]-gingerol’s (10G) anticancer properties clinical trials have revealed that daily consumption of a dose
against human colorectal cancer cells stem from its ability to of up to 5 grams of ginger extract (dry form) are nontoxic
induce slow and sustained release of calcium ions from en- with the exception of mild stomach upset in individuals who
doplasmic reticulum in a concentration-dependent manner do not normally consume spicy foods while higher doses
[45]. Further, 6S was demonstrated to induce cell death in col- (6 grams or more) resulted in gastric irritation with loss of
orectal cancer cells by mechanisms involving modulation of protective intestinal mucosa [54]. In addition to possessing
mitochondrial function though generation of reactive oxygen anticancer properties, ginger constituents are also promising
species (ROS) leading to cytochrome c release [46]. Several candidates for chemopreventive interventions. In this regard,
other studies have confirmed that 6G and 6S induce caspase- topical application of GE has been reported to significantly
dependent apoptosis in colon and prostate cancer cell lines reduce 12-0-tetradecanoylphorbol-13-acetate induced hyper-
through their effects on ERK1/2/JNK/AP-1, STAT3, and NF- plasia and tumor incidence in SENCAR mouse [58]. Simi-
␬B signaling pathways [47, 48]. In another study, a cysteine- larly, zerumbone, a ginger sesquiterpene, has been shown
conjugated metabolite of 6S (M2) was found to be as effective to inhibit 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone
as 6S in inducing apoptotic pathway leading to reduction in induced lung tumorigenesis in A/J mouse [59].
cancer cell proliferation and tumor regression in mouse mod- Although a body of literature supports the preventive and
els but with reduced toxicity against normal cells than 6S [49]. therapeutic antitumor roles of ginger and its constituents,
Another constituent of ginger, 6-dehydrogingerdione (6DG) there is still a long way to go before ginger can be imple-
has been observed to trigger mitochondrial apoptotic path- mented in cancer therapy in humans. There are a num-
way in breast cancer cells via mechanisms involving ROS ber of factors that limit the potential use of ginger to treat
and c-Jun N-terminal kinase [50]. Further, 6DG has been cancer but the most significant ones include poor bioavail-
demonstrated to mediate its anti-inflammatory effects by at- ability of ginger constituents, lack of standard GE for treat-
tenuating enzymes nitric oxide synthase and cyclooxygenase- ment owing to variation in its composition by various factors,
2 and pro-inflammatory cytokines such as interleukin-1␤, and poor understanding of pharmacokinetic properties and
interleukin-6, and tumor necrosis factor-␣ [51]. Ginger phy- metabolic transformations of ginger constituents alone and
tochemicals not only inhibit proliferation of cancer cells but in combination with other components. Further, the max-
can also potentially prevent cancer metastasis by preventing imum levels of 6G achievable in the plasma reported by
invasion of healthy normal cells by cancer cells. To this ef- pharmacokinetic studies are much lower than the reported
fect, [4]-shogaol (4S) has been shown to cause mesenchymal- in vitro effective half-maximal dose thus limiting its poten-
epithelial transition therefore reducing metastasis in breast tial efficacy in humans [57]. Oral bioavailability of various
cancer in animal models [52]. Further, [6]-shogaol has been ginger constituents can vary and depends on multiple fac-
reported to inhibit cancer cell invasion by downregulating tors such as solubility and stability in gastrointestinal fluid,
matrix metalloproteinase-9 in MDA-MB-231 cells [53]. Since membrane permeability, transporter-driven intestinal efflux,
different constituents of ginger exert their effects by targeting presystemic gut wall metabolism, and presystemic hepatic
multiple and diverse pathways, studies conducted with GE, metabolism [60]. Research aimed at devising strategies to
rather than individual components, offer advantage of ex- increase bioavailability of ginger constituents would aid in
ploiting anti-cancer synergy between different components. including ginger in chemotherapeutic regimens. As men-
It has been demonstrated that the combination of ginger phy- tioned earlier, composition of GE depends on various fac-
tochemicals with GE is more effective in inhibiting growth of tors some of which are not under the control of researchers
prostate cancer cells in comparison to isolated phytochemi- such as geographical and environmental factors. Since phar-
cals or GE alone [54]. In a related study, GE was found to be macological effects of ginger depend on its composition,
more effective in inhibiting prostate tumor growth in nude the use of nonstandardized extract, and lack of pharmacoki-
mice than an artificially formulated mix that consisted of the netic signature of GE associated with pharmacological ef-
most abundant components of GE (6G, 8G, 10G, and 6S) fects in different studies limit the use of these results in
[55]. While these synergistic interactions have been amply designing and implementing ginger-based regimens for can-
demonstrated, most of the molecular details pertaining to cer treatment. In view of these facts, it is vital to first es-
their mechanistic interplay are yet to be determined. In one tablish the most effective and standard composition of GE
study based in Japan, researchers tested the efficacy of (GE) for its anticancer effects to increase the reliability and repro-
against pancreatic cancer cell lines. The study demonstrated ducibility of various studies conducted with it. In addition to
that treatment of pancreatic cancer cells with ginger extract this, studies aimed at understanding pharmacokinetic prop-
resulted in inhibition of cell cycle progression with conse- erties and metabolic transformation of ginger constituents
quent induction of ROS-mediated cell death called autosis alone and in combination with other constituents are


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2016, 60, 1364–1373 1369

Figure 2. Benefits of combining gin-


ger with chemotherapeutic regimens in
practice. Various bioactive molecules
present in ginger (shown in fig-
ure) are responsible for its anti-
cancer, antiemetic, antioxidant, and
anti-inflammatory effects. These phys-
iologically important attributes of
ginger can be exploited to allevi-
ate chemotherapy-induced side ef-
fects and to make therapy more ef-
ficacious when used in combination
with chemotherapeutic regimens in
practice.

warranted to better understand the synergy between different can reduce chemotherapy generated gastrointestinal symp-
components. toms such as vomiting and nausea. This would further help in
reducing electrolyte imbalances and severe dehydration and
in regaining lost appetite caused by chemotherapy-induced
5 Combining ginger and chemotherapy: vomiting and nausea. In addition, treatment with chemother-
Taming of the shrew apeutic agents can cause oxidative injury to normal healthy
cells, which can be ameliorated by antioxidant properties of
Chemotherapy is one of the most prevalent interventions various ginger constituents. For instance, 6G has been shown
to treat cancer but it inflicts severe distress to patients ow- to provide cardioprotection against doxorubicin-induced car-
ing to its adverse side effects, which limit its full utiliza- diotoxicity through its antioxidant effects and modulation
tion in treating cancer. Therefore, there is a pressing need of NF-␬B signaling pathway in rat models [61]. It has been
for the development and implementation of alternative ther- proposed that ginger phytochemicals (6G, 8G, 10G and 6S)
apies with mild or no side effects. One strategy that has function as 5-HT3 and NK-1 antagonists [62]. As mentioned
tremendous potential is combining chemotherapy with di- previously, antagonism of 5-HT3 and NK-1 receptors lead to
etary agents such as ginger. This combinatorial approach antiemetic effects by stalling stimulation of vomiting cen-
provides twofold advantage over conventional therapies in ter. Therefore, use of ginger would be as effective as 5-HT3
practice (Fig. 2). First, antiemetic, antioxidant, and anti- and NK-1 receptors antagonists in alleviating chemotherapy-
inflammatory attributes of ginger can be exploited to alleviate induced nausea and vomiting but without the side effects.
chemotherapy-induced side effects. Second, ginger and its Furthermore, anti-inflammatory effects of ginger have been
constituents possess anticancer properties, which can work shown to reduce cell injury in the gastrointestinal tract, which
in synergy with chemotherapy making the combination ther- promotes localized tissue damage through the release of
apy more effective than chemotherapy alone. Here, we em- pro-inflammatory factors [63]. In addition, since ginger con-
phasize on the possibility of using ginger in combination stituents target many signaling pathways to inhibit growth
with chemotherapy with the hope of increasing the effective- of cancer of cells, use of ginger in combination with other
ness of chemotherapy along with ameliorating its side ef- chemotherapeutic agents may result in synergism of their an-
fects. The effectiveness of this combination therapy relies on ticancer effects making combination therapy more effective
antiemetic, antioxidant, and anticancer properties of ginger. than chemotherapy alone. This could possibly reduce the rec-
The antiemetic attributes of bioactive constituents of ginger ommended dose of treatment that would further reduce side


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
1370 R. Saxena et al. Mol. Nutr. Food Res. 2016, 60, 1364–1373

effects. Therefore, the twofold advantage is that ginger can conducted by Zick et al. reported that ginger provides no ad-
be used to prevent/treat cancer development through its ef- ditional benefit to reduce the severity or prevalence of CINV
fects on oxidative stress and inflammation (two major tumor when given with antiemetic agents [69]. Although majority of
initiators and promoters) while subduing the most prevalent studies confirm the effectiveness of ginger in reducing CINV,
side effects of chemotherapy such as vomiting and diarrhea, clinical applicability of these studies is limited due to caveats
which often result in discontinuation of the treatment [64]. associated with them and the existence of conflicting reports.
Although there exists no clinical trial that verified the
efficacy of ginger in treating cancer in humans, there has
been one clinical trial that investigated the role of gin- 6 Future of ginger-augmented
ger extract in chemoprevention of colorectal cancer [65]. chemotherapy: It is looking good
In this pilot study, 21 subjects with high risk for colorec-
tal cancer were assigned to placebo or ginger group, ad- With the identification of the role of dietary and lifestyle
ministered with 2.0 g ginger daily for 28 days and base- factors in cancer pathogenesis, disease prevention and
line and 28-day levels of prostaglandin E2, leukotriene B4, treatment should be directed toward combining beneficial
13-hydroxy-octadecadienoic acids, and 5-, 12-, & 15-hydroxy- dietary agents obtained from fruits and vegetables. The phyto-
eicosatetraenoic acid were quantified in the colonic mucosa chemicals found in fruits and vegetables include carotenoids,
of the subjects. The study reported that ginger was not effec- polyphenolics, anthocyanins, terpenes, and alkaloids which
tive in decreasing eicosanoid levels in subjects at high risk have been reported to target signaling cascades which are
for colorectal cancer [65]. However, it should be noted that specifically altered in cancer cells [70]. Specifically, these phy-
the study was a pilot study associated with limitations such tochemicals are known to prevent DNA damage, reduce ox-
as small sample size and large subject-to-subject variation in idative stress, and regulate inflammatory processes thereby
eicosanoid levels. Therefore, detailed and more systematic reducing the overall risk of cancer development and progres-
clinical trials are warranted to completely explore the chemo- sion [70]. With recent reports suggesting that only less than
preventive and chemotherapeutic roles of ginger. The focus 15% of US adults consume enough fruits and vegetables to
of most ginger-based clinical trials has been toward testing meet their daily recommendations, it is essential to exploit
the effectiveness of ginger in ameliorating chemotherapy- other sources of dietary phenolics for cancer prevention and
induced side effects but no consensus has been achieved on treatment. Ginger qualifies as an excellent candidate to be ap-
this due to a variety of caveats associated with these stud- plied for such interventions and can be used in various ways.
ies. For instance, a randomized, double-blind and placebo- Presence of numerous anticancer agents in ginger points
controlled clinical trial was conducted with 576 patients to as- out that ginger alone can be potentially used to treat can-
sess the effectiveness of ginger in ameliorating acute CIN in cer. However, due to long treatment time associated with any
cancer patients receiving 5-HT3 receptor antagonist antiemet- natural therapy, less aggressive and early stage cancers with
ics [66]. Patients were randomly assigned to one of four long latency are most likely to be benefitted from such ther-
groups: placebo, 0.5, 1.0, or 1.5 g of ginger and were ad- apy. In such a scenario, combining ginger with other dietary
ministered their assigned dosage of ginger or placebo twice phenolics and a chemotherapeutic agent would yield more
daily for a total of 6 days starting 3 days before the first day beneficial results against aggressive and advanced stage can-
of chemotherapy. Severity of nausea was evaluated based on cers. Several dietary agents including grape seed proantho-
a 7-point rating scale (“1” = “Not at all Nauseated” and “7” = cyanidins, green tea polyphenols, silymarin, genistein, and
“Extremely Nauseated”) as reported by patients for days 1–4 curcumin affect cell cycle progression and induce apoptosis
of each cycle. Results of the study showed that daily admin- in various cancer cells [71]. Clinical studies should be aimed
istration of 0.5 and 1.0 g ginger was most effective in reduc- at combining several dietary agents to increase their potency
ing acute chemotherapy-induced nausea. While the major and effectiveness since single agent intervention often fails
strengths of the study include large sample size and double- in disease management due to the development of resistance
blinded treatment to eliminate bias in result evaluation, the with their extended use [72]. To this effect, Rhazya stricta in
major caveat of the study was not controlling for chemother- combination with GE was found to synergistically suppress
apy regimens (i.e., high versus low emetogenic regimens) proliferation and induce apoptosis in human glioblastoma
[66]. Similar beneficial effects of ginger in reducing the preva- cells [73]. This study is an example of how these phytochem-
lence of acute nausea was reported in the study conducted by icals can be synergistically employed to simultaneously tar-
Panahi et al. wherein women with advanced stage breast can- get several characteristic signaling pathways of cancer cells.
cer received daily dose of 1.5 g of ginger in addition to stan- Further, studies aimed at understanding the mechanisms re-
dard antiemetic therapy [67]. Further, a study conducted by sponsible for synergy between ginger and other phytochem-
Pillai et al. confirmed the ability of ginger to reduce acute and icals and chemotherapeutic agents would provide a neces-
delayed CINV in children and young adults receiving high sary framework for the development of ginger-based cancer
emetogenic chemotherapy [68]. In addition to this, there have interventions.
been some contradicting reports that do not support benefits Potential use of ginger in cancer therapy first demands val-
of ginger to treat chemotherapy-associated side effects. Study idation of its anticancer effects in humans. However, use of


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2016, 60, 1364–1373 1371

nonstandard GE in studies related to its anticancer properties [7] Nichenametla, S. N., Taruscio, T. G., Barney, D. L., Exon, J.
and lack of knowledge of pharmacokinetic and pharmacody- H., A review of the effects and mechanisms of polyphenolics
namic relationship for its constituents represent major obsta- in cancer. Crit. Rev. Food Sci. Nutr. 2006, 46, 161–183.
cles in clinical translation of these studies. Pharmacological [8] Haniadka, R., Saldanha, E., Sunita, V., Palatty, P. L. et al.,
effects of ginger depend on its constituents such as gingerols, A review of the gastroprotective effects of ginger (Zingiber
shogaols, and paradols whose concentrations in ginger de- officinale roscoe). Food Funct. 2013, 4, 845–55.
pends on many factors. In addition, compositional variations [9] Carr, C., Ng, C. Wigmore, T., The side effects of chemothera-
in different GE preparations can arise from different harvest peutic agents. Curr. Anaesth. Crit. Care 2008, 19, 70–79.
times and methods of harvest, storage, and extraction. The [10] Abal, M., Andreu, J. M., Barasoain, I., Taxanes: microtubule
foremost step toward practical application of ginger in cancer and centrosome targets, and cell cycle dependent mecha-
therapy is to identify the most effective ginger composition as nisms of action. Curr. Cancer Drug Targets 2003, 3, 193–203.
a standard extract which can be universally used for ginger- [11] Emadi, A., Jones, R.J., Brodsky, R. A., Cyclophosphamide
based studies. One potential strategy to standardize extract and cancer: golden anniversary. Nat. Rev. Clin. Oncol. 2009,
preparations is to establish a compositional fingerprint cor- 6, 638–647.
responding to most efficacious composition of ginger. This [12] Florea, A. M., Büsselberg, D., Cisplatin as an anti-tumor drug:
fingerprint can serve as gold standard for extract preparations cellular mechanisms of activity, drug resistance and induced
for ginger-based studies worldwide. Results obtained from side effects. Cancers 2011, 3, 1351–1371.
these studies would be much more reliable and can be conve- [13] Rothenberg, M. L., Irinotecan (CPT-11): Recent develop-
niently compared with each other to draw holistic inferences ments and future directions-colorectal cancer and beyond.
on biological effects of ginger. The use of alternative therapies Oncologist 2001, 6, 66–80.
has gained popularity in the recent years with dependence on [14] Ogden, A., Rida, P.C., Reid, M. D., Aneja, R., Interphase micro-
chemopreventive and chemotherapeutic dietary agents to re- tubules: chief casualties in the war on cancer? Drug Discov.
duce toxicity and resistance encountered with current clinical Today 2014, 19, 824–829.
drugs [4, 72]. The future of milder and more effective cancer [15] Di Fiore, F., Van Cutsem, E., Acute and long-term gastroin-
therapy relies on studies and clinical trials conducted with testinal consequences of chemotherapy. Best Pract. Res.
systematic and regimented combinations of dietary pheno- Clin. Gastroenterol. 2009, 23, 113–24.
lics alone and in combination with chemotherapeutic agents [16] Herrstedt, J., Dombernowsky, P., Anti-emetic therapy in can-
in practice. The low-cost and down-to-earth strategy of incor- cer chemotherapy: current status. Basic Clin. Pharmacol.
porating the ginger rhizome and other dietary phenolics in Toxicol. 2007, 101, 143–150.
routine cancer therapy could thus potentially make a substan- [17] Jordan, K., Kasper, C., Schmoll, H. J., Chemotherapy-
tial difference to cancer patients’ outcomes and mitigate the induced nausea and vomiting: current and new standards
toll that chemotherapy takes, naturally. in the antiemetic prophylaxis and treatment. Eur. J. Cancer
2005, 41, 199–205.
The authors would like to acknowledge Angela Ogden for her
[18] Hesketh, P. J., Kris, M. G., Grunberg, S. M., Beck, T. et al.,
input in designing the figures. Proposal for classifying the acute emetogenicity of cancer
The authors have declared no conflict of interest. chemotherapy. J. Clin. Oncol. 1997, 15, 103–109.
[19] Minami, M., Endo, T., Hirafuji, M., Hamaue, N. et al., Pharma-
cological aspects of anticancer drug-induced emesis with
emphasis on serotonin release and vagal nerve activity.
7 References Pharmacol. Ther. 2003, 99, 149–165.
[20] Berger, M., Gray, J. A., Roth, B. L., The expanded biology of
[1] IMS: Global Oncology Trend Report serotonin. Annu. Rev. Med. 2009, 60, 355–366.
[2] Hanahan, D., Weinberg, R. A., Hallmarks of cancer: the next [21] Navari, R. M., Management of chemotherapy-induced nau-
generation. Cell 2011, 144, 646–674. sea and vomiting: focus on newer agents and new uses for
[3] Anand, P., Kunnumakkara, A. B., Sundaram, C., Harikumar, older agents. Drugs 2013, 73, 249–262.
K.B. et al., Cancer is a preventable disease that requires ma- [22] Rao, K. V., Faso, A., Chemotherapy-induced nausea and vom-
jor lifestyle changes. Pharm. Res. 2008, 25, 2097–2116. iting: optimizing prevention and management. Am. Health
[4] Steward, W. P., Brown, K., Cancer chemoprevention: a Drug Benefits 2012, 5, 232–240.
rapidly evolving field. Br. J. Cancer 2013, 109, 1–7. [23] Saliba, F., Hagipantelli, R., Misset, J. L., Bastian, G. et al.,
[5] Wilson, K. M., Giovannucci, E. L., Mucci, L. A., Lifestyle and Pathophysiology and therapy of irinotecan-induced delayed-
dietary factors in the prevention of lethal prostate cancer. onset diarrhea in patients with advanced colorectal cancer:
Asian J. Androl. 2012, 14, 365–374. a prospective assessment. J. Clin. Oncol. 1998, 16, 2745–
2751.
[6] Vahid, F., Zand, H., Nosrat-Mirshekarlou, E., Najafi, R. et al.,
The role dietary of bioactive compounds on the regulation of [24] Ikuno, N., Soda, H., Watanabe, M., Oka, M., Irinotecan (CPT-
histone acetylases and deacetylases: a review. Gene 2015, 11) and characteristic mucosal changes in the mouse ileum
562, 8–15. and cecum. J. Natl. Cancer Inst. 1995, 87, 1876–1883.


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
1372 R. Saxena et al. Mol. Nutr. Food Res. 2016, 60, 1364–1373

[25] Meyersohn, N. M., Pursnani, A., Neilan, T. G., Detection of [42] Kim, S. O., Kundu, J. K., Shin, Y. K., Park, J. H. et al., [6]-
cardiac toxicity due to cancer treatment: role of cardiac MRI. Gingerol inhibits COX-2 expression by blocking the activa-
Curr. Treat. Options Cardiovasc. Med. 2015, 17, 1–9. tion of p38 MAP kinase and NF-kappaB in phorbol ester-
[26] Dietrich, J., Prust, M., Kaiser, J., Chemotherapy, cognitive stimulated mouse skin. Oncogene 2005, 24, 2558–2567.
impairment and hippocampal toxicity. Neuroscience 2015, [43] Ahn, S. I., Lee, J. K., Youn, H. S., Inhibition of homodimeriza-
doi:10.1016/j.neuroscience.2015.06.016. tion of toll-like receptor 4 by 6-shogaol. Mol. Cells 2009, 27,
[27] Joly, F., Rigal, O., Noal, S., Giffard, B., Cognitive dysfunction 211–215.
and cancer: which consequences in terms of disease man- [44] Lee, S. H., Cekanova, M., Baek, S. J., Multiple mechanisms
agement. Psycho-Oncology 2011, 12, 1251–1258. are involved in 6-gingerol-induced cell growth arrest and
[28] Govindarajan, V. S., Ginger: chemistry, technology, and qual- apoptosis in human colorectal cancer cells. Mol. Carcinog.
ity evaluation: part 1. Crit. Rev. Food Sci. Nutr. 1982a, 17, 2008, 47, 197–208.
1–96. [45] Chen, C. Y., Li, Y. W., Kuo, S. Y., Effect of [10]-gingerol
[29] Govindarajan, V.S., Ginger: chemistry, technology, and qual- on [Ca2+ ]i and cell death in human colorectal cancer cells.
ity evaluation: part 2. Crit. Rev. Food Sci. Nutr. 1982b 17, Molecules 2009, 14, 959–969.
189–258. [46] Pan, M. H., Hsieh, M. C., Kuo, J. M., Lai, C. S. et al., 6-Shogaol
[30] Vasala, P. A., in: Peter, K. V. (Ed.), Handbook of Herbs and induces apoptosis in human colorectal carcinoma cells via
Spices, Vol. 1, Woodhead Publishing, Cambridge, UK 2004, ROS production, caspase activation, and GADD 153 expres-
pp. 195–206. sion. Mol. Nutr. Food Res. 2008, 52, 527–537.

[31] Zick, S. M., Djuric, Z., Ruffin, M. T, Litzinger, A. J. et al., [47] Radhakrishnan, E. K., Bava, S. V., Narayanan, S.S., Nath,
Pharmacokinetics of 6-gingerol, 8-gingerol, 10-gingerol, and L.R., [6]-Gingerol induces caspase-dependent apoptosis and
6-shogaol and conjugate metabolites in healthy human sub- prevents PMA-induced proliferation in colon cancer cells by
jects. Cancer Epidemiol. Biomarkers Prev. 2008, 17, 1930– inhibiting MAPK/AP-1 signaling. PLOS One 2014, 9, e104401.
1936. [48] Saha, A., Blando, J., Silver, E., Beltran, L. et al., 6-Shogaol
[32] Schieber, M., Chandel, N. S., ROS function in redox signaling from dried ginger inhibits growth of prostate cancer cells
and oxidative stress. Curr. Biol. 2014, 24, R453–R462. both in vitro and in vivo through inhibition of STAT3 and
NF-␬B signaling. Cancer Prev. Res. 2014, 7, 627–638.
[33] Baliga, M. S., Haniadka, R., Pereira, M. M., D’Souza, J. J.
et al., Update on the chemopreventive effects of ginger and [49] Warin, R. F., Chen, H., Soroka, D. N., Zhu, Y. et al., Induction
its phytochemicals. Crit. Rev. Food Sci. Nutr. 2011, 51, 499– of lung cancer cell apoptosis through a p53 Pathway by [6]-
523. shogaol and its cysteine-conjugated metabolite M2. J. Agric.
Food Chem. 2014, 62, 1352–1362.
[34] Rahmani, A. H., Shabrmi, F. M., Aly, S. M., Active ingredi-
ents of ginger as potential candidates in the prevention and [50] Hsu, Y. L., Chen, C. Y., Hou, M. F., Tsai, E. M. et al., 6-
treatment of diseases via modulation of biological activities. Dehydrogingerdione, an active constituent of dietary gin-
Int. J. Physiol. Pathophysiol. Pharmacol. 2014, 6, 25–36. ger, induces cell cycle arrest and apoptosis through reactive
oxygen species/c-Jun N-terminal kinase pathways in human
[35] Grzanna, R., Lindmark, L., Frondoz, C. G., Ginger-An herbal
breast cancer cells. Mol. Nutr. Food Res. 2010, 54, 1307–1317.
medicinal product with broad anti-inflammatory actions. J.
Med. Food 2005, 8, 125–132. [51] Huang, S. H., Lee, C. H., Wang, H. M., Chang, Y. W. et al.,
6-Dehydrogingerdione restrains lipopolysaccharide-induced
[36] Tjendraputra, E., Tran, V. H., Liu-Brennan, D., Roufogalis, B.
inflammatory responses in RAW 264.7 macrophages. J.
D. et al., Effect of ginger constituents and synthetic ana-
Agric. Food Chem. 2014, 62, 9171–9179.
logues on cyclooxygenase-2 enzyme in intact cells. Bioorg.
Chem. 2001, 29, 156–163. [52] Hsu, Y. L., Chen, C. Y., Lin, I. P., Tsai, E. M. et al., 4-Shogaol,
an active constituent of dietary ginger, inhibits metastasis
[37] Flynn, D. L., Rafferty, M. F., Inhibition of human neurotrophil
of MDA-MB-231 human breast adenocarcinoma cells by de-
5-lipoxygenase activity by gingerdione, shagaol, capsaicin
creasing the repression of NF-␬B/Snail on RKIP. J. Agric.
and related pungent compounds. Prostaglandins Leukot.
Food Chem. 2012, 60, 852–861.
Med. 1986, 24, 195–198.
[53] Ling, H., Yang, H., Tan, S.H., Chui, W. K. et al., 6-Shogaol,
[38] Laufer, S., Role of eicosanoids in structural degradation in
an active constituent of ginger, inhibits breast cancer cell
osteoarthritis. Curr. Opin. Rheumatol. 2003, 15, 623–627.
invasion by reducing matrix metalloproteinase-9 expression
[39] Lawrence, T., The nuclear factor NF-␬B pathway in inflam- via blockade of nuclear factor-␬B activation. Br. J. Pharmacol.
mation. Cold Spring Harb. Perspect. Biol. 2009, 1, a001651. 2010, 161, 1763–1777.
[40] Frondoza, C. G., Sohrabi, A., Polotsky, A., Phan, P. V. et al., [54] Brahmbhatt, M., Gundala, S. R., Asif, G., Shamsi, S. A. et al.,
An in vitro screening assay for inhibitors of proinflamma- Ginger phytochemicals exhibit synergy to inhibit prostate
tory mediators in herbal extracts using human synoviocyte cancer cell proliferation. Nutr. Cancer 2013, 65, 263–272.
cultures. In vitro 2004, 30, 95–101.
[55] Gundala, S.R., Mukkavilli, R., Yang, C., Yadav, P. et al., En-
[41] Kim, S. O., Chun, K. S., Kundu, J. K., Surh, Y. J., Inhibitory terohepatic recirculation of bioactive ginger phytochemi-
effects of [6]-gingerol on PMA-induced COX-2 expression cals is associated with enhanced tumor growth-inhibitory
and activation of NF-kappaB and p38 MAPK in mouse skin. activity of ginger extract. Carcinogenesis 2014, 35, 1320–
Biofactors 2004, 21, 27–31. 1329.


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com
Mol. Nutr. Food Res. 2016, 60, 1364–1373 1373

[56] Akimoto, M., Iizuka, M., Kanematsu, R., Yoshida, M. et al., [68] Pillai, A. K., Sharma, K. K., Gupta, Y. K., Bakhshi, S. et al.,
Anticancer effect of ginger extract against pancreatic can- Anti-emetic effect of ginger powder versus placebo as an
cer cells mainly through reactive oxygen species-mediated add-on therapy in children and young adults receiving high
autotic cell death. PLOS One 2015, 10, e0126605. emetogenic chemotherapy. Pediatr. Blood Cancer 2011, 56,
[57] Karna, P., Chagani, S., Gundala, S. R., Rida, P. C. et al., Benefits 234–238.
of whole ginger extract in prostate cancer. Br. J. Nutr. 2012, [69] Zick, S. M., Ruffin, M. T., Lee, J., Normolle, D. P. et al.,
107, 473–484. Phase II trial of encapsulated ginger as a treatment for
[58] Katiyar, S. K., Agarwal, R., Mukhtar, H., Inhibition of tumor chemotherapy-induced nausea and vomiting. Support Care
promotion in SENCAR mouse skin by ethanol extract of Zin- Cancer 2009, 17, 563–572.
giber officinale rhizome. Cancer Res. 1996, 56, 1023–1030. [70] Banudevi, S., Swaminathan, S., Maheswari, K. U., Pleiotropic
[59] Kim, M., Miyamoto, S., Yasui, Y., Oyama, T. et al., Zerumbone, role of dietary phytochemicals in cancer: emerging perspec-
a tropical ginger sesquiterpene, inhibits colon and lung car- tives for combinational therapy. Nutr. Cancer 2015, 67, 1021–
cinogenesis in mice. Int. J. Cancer 2009, 124, 264–271. 1048.

[60] Mukkavilli, R., Gundala, S.R., Yang, C., Donthamsetty, S. [71] Singh, R. P., Dhanalakshmi, S., Agarwal, R., Phytochemicals
et al., Modulation of cytochrome P450 metabolism and trans- as cell cycle modulators-a less toxic approach in halting hu-
port across intestinal epithelial barrier by ginger biopheno- man cancers. Cell Cycle 2002, 1, 156–161.
lics. PLOS One 2014, 9, e108386. [72] Sak, K., Chemotherapy and dietary phytochemi-
[61] El-Bakly, W. M., Louka, M. L., El-Halawany, A. M., Schaalan, cal agents. Chemother. Res. Pract. 2012, doi:10.1155
M. F., 6-gingerol ameliorated doxorubicin-induced cardiotox- /2012/2825702012;2012:282570.
icity: role of nuclear factor kappa B and protein glycation. [73] Elkady, A. I., Hussein, R. A., Abu-Zinadah, O. A., Effects
Cancer Chemother. Pharmacol. 2012, 70, 833–841. of crude extracts from medicinal herbs Rhazya stricta and
[62] Haniadka, R., Rajeev, A. G., Palatty, P. L., Arora, R. et al., Zingiber officinale on growth and proliferation of human
Zingiber officinale (ginger) as an anti-emetic in cancer brain cancer cell line in vitro. Biomed. Res. Int. 2014,
chemotherapy: a review. J. Altern. Complement. Med. 2012, doi:10.1155/2014/260210.
18, 440–44. [74] Endo, T., Minami, M., Monma, Y., Yoshioka, M. et al., Effect
[63] Marx, W., Ried, K., McCarthy, A. L., Vitetta, L. et al., of GR38032F on cisplatin and cyclophosphamide-induced
Ginger-mechanism of action in chemotherapy-induced nau- emesis in the ferret. Biog. Amines 1990, 7, 525-533.
sea and vomiting: a review. Crit. Rev. Food Sci. Nutr. 2015, [75] Endo, T., Minami, M., Monma, Y., Yoshioka, M. et al., Vago-
doi:10.1080/10408398.2013.865590. tomy and ondansetron (5-HT3 antagonist) inhibited the
[64] Arbuckle, R. B., Huber, S. L., Zacker, C., The consequences increase of serotonin concentration induced by cytotoxic
of diarrhea occurring during chemotherapy for colorec- drugs in the area postrema of ferrets. Biog. Amines 1992,
tal cancer: a retrospective study. Oncologist 2000, 5, 250– 9, 163–175.
259. [76] Manju, V., Nalini, L., Chemopreventive efficacy of gin-
[65] Zick, S. M., Turgeon, D. K., Ren, J., Ruffin, M. T. et al., Pilot clin- ger, a naturally occurring anticarcinogen during the initia-
ical study of the effects of ginger root extract on eicosanoids tion, post-initiation stages of 1,2 dimethylhydrazine-induced
in colonic mucosa of subjects at increased risk for colorectal colon cancer. Clin. Chim. Acta 2005, 358, 60–67.
cancer. Mol. Carcinog. 2015, 54, 908–915. [77] Hung, J. Y., Hsu, Y. L., Li, C. T., Ko, Y. C. et al., 6-shogaol,
[66] Ryan, J. L., Heckler, C. E., Roscoe, J. A., Dakhil, S. R. et al., an active constituent of dietary ginger, induces autophagy
Ginger (Zingiber officinale) reduces acute chemotherapy- by inhibiting the AKT/mTOR pathway in human non-small
induced nausea: a URCC CCOP study of 576 patients. Sup- cell lung cancer A549 cells. J. Agric. Food Chem. 2009, 57,
port Care Cancer, 2012, 20, 1479–1489. 9809–9816.

[67] Panahi, Y., Saadat, A., Sahebkar, A., Hashemian, F. et al., [78] Brown, A. C., Shah, C., Liu, J., Pham, J. T. et al., Ginger’s
Effect of ginger on acute and delayed chemotherapy-induced (Zingiber officinale Roscoe) inhibition of rat colonic adeno-
nausea and vomiting: a pilot, randomized, open-label clinical carcinoma cells proliferation and angiogenesis in vitro. Phy-
trial. Integr. Cancer Ther. 2012, 11, 204–211. tother. Res. 2008, 23, 640–645.


C 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.mnf-journal.com

You might also like